MicroRNA-377 predicts poor clinical outcome of gastric cancer and induces tumorigenesis by targeting multiple tumor-suppressor genes

  • Authors:
    • Xu Wen
    • Jian-Qiu Wu
    • Wei Peng
    • Ji-Feng Feng
    • Jin-Hai Tang
  • View Affiliations

  • Published online on: May 15, 2015     https://doi.org/10.3892/or.2015.3981
  • Pages: 203-210
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is a major cause of cancer mortality worldwide. MicroRNAs are evolutionally conserved small non-coding RNAs that are critical for the regulation of gene expression. The aberrant expression of microRNA (miRNA) is involved in tumorigenesis and prognosis. In the present study, the clinical significance of miR-377 was assessed using RT‑qPCR and MTT assay. The results showed that the expression of miR-377 was upregulated in GC compared with normal gastric tissues, and its expression level was increased in GC cell lines compared with normal gastric cells. In addition, there was a significant association between miR-377 expression and clinicopathological characteristics, in particular distant metastasis, TNM stage and early recurrence. GC patients with a higher miR-377 expression showed significantly poorer overall survival (OR) and shorter time to recurrence than those with a lower miR-377 expression. The Cox regression analysis identified miR-377 overexpression as an independent prognostic factor for GC. Overexpression of miR-377 in MKN-45 GC cells significantly promoted cell proliferation, whereas the suppression of miR-377 inhibited these effects. Furthermore, miR-377 downregulated p53, PTEN and TIMP1 expression by directly targeting the 3'-untranslated region of these target genes. Collectively, miR-377 potentially served as a new molecular predictive biomarker of GC tumorigenesis and prognosis, which may be useful in targeted therapy and the prognosis of GC patients.

Introduction

Gastric cancer (GC) remains the fourth most commonly diagnosed cancer and is the second leading cause of cancer-associated mortality worldwide (1). The carcinogenesis of GC is complicated and involves the dysregulation of oncogenes and tumor suppressors (2). Many molecules are responsible for GC tumorigenesis, and despite advances in surgical and chemotherapeutic interventions, the overall 5-year survival for GC patients remains low (30%) and the recurrence rate high (35). Thus, identifying new biomarkers or factors that improve the diagnosis of GC prior to tumorigenesis and predict prognosis responsible for cancer recurrence in patients with GC is crucial.

A number of microRNA (miRNA) genes have been identified in the human genome and it has been suggested that at least 50% of all protein-encoding genes are regulated by miRNA (6). Mature miRNAs are 21–22 nucleotides in size and affect the post-translational expression of genes by interacting with complementary target sites within the 3′-untranslated region of the messenger RNA (7). Recent findings have shown the role of miRNAs in a variety of basic biological and pathological processes (8) and the association of miRNA signatures with human diseases has been established (9). miR-377 can have a critical role in the pathophysiology of diabetic nephropathy (10) and human nucleus pulposus cells (11). miR-377 was responsible for metabolizing the excess heme generated during hemolysis (12). miR-377 may be required for glioblastoma multiforme (GBM) development and serve as a therapeutic target for the treatment of GBM (13). However, the expression of miR-377 in GC and its prognostic values remain to be elucidated.

In the present study, we evaluated the clinical significance of miR-377. The expression level of miR-377 was overexpressed in GC tissues and cell lines. Furthermore, the correlation between the expression level of miR-377 and clinicopathological characteristics was analyzed. The prognostic value of miR-377 on the prognosis of GC patients was also estimated. Further analyses showed that the overexpression of miR-377 promoted GC cell proliferation. Moreover, p53, PTEN and TIMP1 were identified as direct targets of miR-377.

Materials and methods

Patients and specimens

This study was approved by the Medical Ethics Committee of Jiangsu Cancer Hospital (Nanjing, China) and written informed consent was obtained from all of the patients. The specimens were handled and made anonymous according to the ethical and legal standards. Paired tissue specimens (tumor and adjacent normal mucosa) from 102 patients with GC were obtained and histologically confirmed by a pathologist at Jiangsu Cancer Hospital and the First People’s Hospital of Yunnan Province from January, 2007 to December, 2010. The samples were derived from patients who had not received adjuvant treatment including radiotherapy or chemotherapy prior to surgery in order to eliminate potential treatment-induced changes to gene expression profiles. After excision, the tissue specimens were immediately frozen in liquid nitrogen for subsequent analysis. The specimens were stained with hematoxylin and eosin and examined histopathologically. The sections that consisted of >80% carcinoma cells were used to prepare the total RNA. Clinicopathological information, including gender, age, tumor size, depth of invasion, lymph node invasion, distant metastasis, TNM stage, tumor differentiation and early recurrence was available for all the patients.

Cell culture

The human HGC-27, MKN-45, BGC-823, AGS, SGC-7901, MGC80-3, NCI-N87 and SNU-1 gastric cancer cell lines, and GES-1 normal gastric cell line were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). MKN-45, BGC-823, AGS, SGC-7901 and SNU-1 cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) and HGC-27, MGC80-3, NCI-N87 and GES-1 were grown in RPMI-1640. In each case, the medium was supplemented with 15% fetal calf serum (FCS), 100 U/ml penicillin and 100 U/ml streptomycin. The cells were incubated at 37°C in a humidified incubator containing 5% CO2.

Reverse transcriptase-quantitative PCR (RT-qPCR)

Approximately 40 mg of tissue sample or harvested cells (2×106) was homogenized in 1 ml TRIzol reagent (Invitrogen, Carlsbad, CA, USA), according to the manufacturer’s instructions. The purity and concentration of RNA were determined using a NanoDrop 1000 spectrophotometer (Thermo Fisher scientific, Wilmington, DE, USA). RNA (2 µg) was reverse transcribed from each sample to produce cDNA using the PrimeScript RT reagent kit (Takara Biotechnology, Dalian, China). Quantitative PCR was employed to determine the relative expression level of target genes using the SYBR Premix ExTaq II kit (Takara) on the 7300 Real-Time PCR systems (Applied Biosystems, Carlsbad, CA, USA). The PCR cycling profile was denatured at 95°C for 30 sec, followed by 40 cycles of annealing at 95°C for 5 sec and extension at 60°C for 34 sec. Small nucleolar RNA U6 was used as an internal standard for normalization. The cycle threshold (CT) value was calculated. The 2−ΔCT (ΔCT =CTmiR-377−CTU6RNA) method was used to quantify the relative amount of miR-377.

miRNA mimics, plasmids and transfection

The miR-377 mimic, miR-377 mutant (miR-377-mut) and miR-377-inhibitor (miR-377-in) were purchased from the shanghai GenePharma, Co. (Shanghai, China), together with the negative control (miR-control). The transfection of miRNAs (50 nM) was performed using X-tremeGENE (Roche, Reinach, Switzerland) according to the manufacturer’s instructions. The 3′-UTR of p53, PTEN and TIMP1 was PCR-amplified from MKN-45 genomic DNA and cloned downstream of the luciferase gene in the pGL vector (Promega, Madison, WI, USA). For the reporter assay, the cells were cultured in 96-well plates and transfected with luciferase reporters (50 ng) and 50 nM of miR-control, miR-377 mimics or miR-377-mut. After 48 h, luciferase activity was measured using the dual-luciferase reporter system (Promega). The Renilla activity was used as an internal control. Each transfection was performed in triplicate.

MTT assay

The cells were seeded in 96-well plates (2×103/well) 24 h after transfection. MTT (Beyotime, Haimen, China) was added to each well and the cells were cultured for 4 h at 37°C. The reaction was stopped by 150 µl DMSO and the optical density at 490 nm was detected on a microplate reader (Thermo Fisher scientific, Kalamazoo, MI, USA).

Colony formation assay

Five hundred of each transfected cells were plated in 6-well plates and cultured for 14 days without any disturbance. The cells were stained with 0.5% crystal violet for 1 h at 37°C. visible colonies were counted in four different fields and the mean value was calculated.

Cell cycle analysis

The cells were seeded into 6-well plates with a density of 1×105 cells/well after transfection and maintained in DMEM/RPMI-1640 containing 15% FCS. Cultured cells were trypsinized after 48 h and fixed with 70% ethanol at 4°C overnight before being stained with propidium iodide (PI) (Invitrogen). DNA contents were detected using an LSRII flow cytometer (BD Biosciences, san Jose, CA, USA). Data were analyzed by Flow Jo (Tree Star, Ashland, OR, USA). TargetScan, miRanda and DIANA were used to determine the putative human protein-coding gene targets of miR-377.

Luciferase reporter assays

MKN-45 cells were co-transfected with miR-377 or control or the mutated 3′-UTR (Mut) of p53, PTEN and TIMP1. After 48 h, the cells were collected and luciferase activity was assayed using the dual-luciferase assay system (Promega).

Protein isolation and western blot analysis

Total proteins were extracted with RIPA lysis buffer with proteinase/phosphatase inhibitors (Beyotime). The lysate was separated by 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis and the gel was blotted onto a PVDF membrane (Bio-Rad, Hercules, CA, USA). The membrane was blocked in 1% BSA and then incubated with one of the following antibodies: anti-p53 (Cellsignal, no. 2527), anti-PTEN (Cellsignal, no. 5384), anti-TIMP1 (Cellsignal, no. 8946) or anti-GAPDH (Cellsignal, no. 2118). Goat radish peroxidase-conjugated anti-rabbit IgG (Cellsignal, no. 2975) were incubated as the secondary antibodies. Subsequent visualization was detected using a SuperSignal West Femto Maximum Sensitivity Substrate (Thermo Fisher scientific).

Statistical analysis

Data are presented as means ± SD. A comparison of the level of miR-377 expression between GC and adjacent normal tissue was performed using the Wilcoxon test. The correlation between the expression of miR-377 and clinicopathological characteristics was assessed using a two-sample Student’s t-test. The postoperative survival rate was analyzed using the Kaplan-Meier method and differences in survival rates were assessed with the log-rank test. A Cox proportional hazards analysis was performed to calculate the hazard ratio (HR) and the 95% confidence interval (CI) to determine the association between miR-377 expression and survival. In addition, a multivariate Cox regression was performed to adjust for other covariates. The tests were two-tailed and results with P<0.05 were considered statistically significant. Statistical analyses were performed using SPSS 19.0 software (SPSS, Chicago, IL, USA).

Results

miR-377 was overexpressed in human GC tissues and cell lines

As shown in Fig. 1A, the overall expression level of miR-377 was determined by RT-qPCR in 102 paired normal and GC tissues. A significantly different expression level of miR-377 between the normal and tumor groups was identified (P<0.0001). Furthermore, miR-377 expression in tumor tissues showed elevated levels of miR-377 compared to the corresponding normal tissues, with an average increase of 9.62-fold. Levels of miR-377 in the BGC-823, AGS, SGC-7901, MGC80-3, NCI-N87 and SNU-1 GC cell lines HGC-27, MKN-45, were also higher than that in the normal GES-1 cell line (P<0.001, Fig. 1B).

Correlations between miR-377 expression and clinicopatho- logical characteristics

To identify the clinical relevance of miR-377 expression in GC, the correlations between miR-377 expression and clinicopathological parameters such as gender, age, tumor size, depth of invasion, lymph node invasion, distant metastasis, TNM stage, tumor differentiation and early recurrence were examined (Table I). Of the 102 GC patients, 82 cases were included in the miR-377-high group and the remaining 20 cases were included in the miR-377-low group. As shown in Table I, the results demonstrated that a high expression of miR-377 was significantly correlated with distant metastasis (P=0.034), TNM stage (P=0.030) and early recurrence (P=0.044). However, there was no significant association between miR-377 expression and other clinicopathological chacteristics, such as gender, age and tumor size, depth of invasion, lymph node invasion and tumor differentiation (P>0.05).

Table I

Clinicopathological parameters of GC patients and the correlation with miR-377 expression.

Table I

Clinicopathological parameters of GC patients and the correlation with miR-377 expression.

CharacteristicsNo.miR-377 expression
P-value
LowHigh
Gender0.328
 Male671552
 Female35530
Age (years)0.936
 <60621250
 ≥6040832
Tumor size (cm)0.174
 <4.5631548
 ≥4.539534
Depth of invasion0.942
 T1,T221417
 T3,T4811665
Lymph node invasion0.085
 Absent781860
 Present24222
Distant metastasis0.034
 Absent401228
 Present62854
TNM stage0.030
 I,II351124
 III,IV67958
Tumor differentiation0.258
 Well, moderate601446
 Poor, mucinous42636
Early recurrence0.044
 No411229
 Yes61853

[i] Bold, statistically significant (P<0.05).

Prognostic value of miR-377

The results showed that miR-377 was a prognostic factor for overall survival (OS) and time to recurrence (TTR) in GC. The Log-rank test results showed that the 3- and 5-year OS rates in the group with miR-377 upregulation were significantly lower than those in the group with miR-377 downregulation (P=0.0106) (Fig. 2A). Similarly, the cumulative TTR rates in the miR-377 upregulation group were significantly higher than those in the miR-377 downregulation group (P=0.0004) (Fig. 2B). Furthermore, the Cox regression analysis suggested that distant metastasis and miR-377 expression status were independent factors that affected OS (P=0.027, HR=1.62, 95% CI=1.13–2.78), while miR-377 expression status was an independent factor for TTR (P=0.022, HR=2.14, 95% CI=0.87-4.42) (Table II). Collectively, these data suggested that miR-377 was an independent prognostic biomarker in GC.

Table II

Univariate and multivariate analyses of prognostic variables of overall survival and time to recurrence in GC patients.

Table II

Univariate and multivariate analyses of prognostic variables of overall survival and time to recurrence in GC patients.

VariablesUnivariate analysis
Multivariate analysis
HR95% CIP-valueHR95% CIP-value
 Overall survival3.563.28–4.340.713
 Gender (male vs. female)2.772.65–3.980.248
 Age (<60 vs. ≥60)2.812.36–3.510.057
 Tumor size (<4.5 vs. ≥4.5)2.151.76–3.010.323
 Depth of invasion (T1,T2 vs. T3,T4)1.881.45–3.540.082
 Lymph node invasion (absent vs. present)2.551.76–4.220.0131.731.26–2.630.091
 Distant metastasis (absent vs. present)1.390.88–3.190.0251.821.04–3.150.016
 TNM stage (I,II vs. III,IV)1.140.63–1.790.0140.960.65–1.610.065
 Tumor differentiation1.371.01–2.630.419
  (Well, moderate vs. poor, mucinous)
Early recurrence (no vs. yes)1.190.88–2.690.0351.651.17–3.050.043
 miR-377 expression (low vs. high)1.481.09–2.430.0061.621.13–2.780.027
Time to recurrence
 Gender (male vs. female)2.081.86–2.830.216
 Age (<60 vs. ≥60)2.682.38–3.590.971
 Tumor size (<4.5 vs. ≥4.5)4.023.21–4.460.317
 Depth of invasion (T1,T2 vs. T3,T4)2.942.62–3.940.462
 Lymph node invasion (absent vs. present)3.272.95–4.090.0132.971.89–3.320.093
 Distant metastasis (absent vs. present)2.380.52–4.250.0453.713.39–4.670.232
 TNM stage (I,II vs. III,IV)1.920.67–4.290.0182.512.19–3.330.307
 Tumor differentiation1.560.18–3.080.054
  (Well, moderate vs. poor, mucinous)
Early recurrence (no vs. yes)1.230.54–2.610.0272.030.17–3.850.041
 miR-377 expression (low vs. high)0.770.41–2.870.0122.140.87–4.420.022

[i] HR, hazard ratio; 95% CI, 95% confidence interval. Bold, statistically significant (P<0.05).

miR-377 promotes GC cell proliferation

To examine the role of miR-377 in GC tumorigenesis, we examined the effect of miR-377 overexpression and inhibition on the proliferation of MKN-45 GC cell lines. The cells were transfected with miR-377 mimic (miR-377), miR-377 inhibitor (miR-377-in) or the miR scramble control oligonucleotides (Control). RT-qPCR showed that miR-377 was significantly increased in the cells transfected with miR-377 mimics and decreased in the miR-377 inhibitor group compared with the control (Fig. 3A). The MTT assay showed that the overexpression of miR-377 significantly promoted the proliferation of MKN-45 cells, whereas the inhibition of miR-377 suppressed cell proliferation (Fig. 3B). The colony formation assay was performed to further confirm the effect of miR-377 on GC cell proliferation, and data indicated that the overexpression of miR-377 significantly increased colony numbers in MKN-45 cell cultures, whereas the knockdown of miR-377 expression obviously decreased colony formation (Fig. 3C). Furthermore, we assessed the cell cycle by flow cytometry. As shown in Fig. 3D, the transfection of miR-377 decreased the percentage of cells in G1 peak but increased that in the s peak. Similarly, miR-377-in led to cell cycle arrests in MKN-45 cells. Collectively, these results showed that miR-377 promotes GC cell growth.

miR-377 directly targets p53, PTEN and TIMP1

To gain insight into the biological implications of miR-377 on GC tumorigenesis, we used TargetScan, miRanda and DIANA for the putative human protein-coding gene targets of miR-377. The tumor-suppressor genes p53, PTEN and TIMP1 were predicted to have miR-377-binding elements in their 3′-UTRs with high fidelity scores (Fig. 4A). To assess whether the predicted miR-377-binding sites in the 3′-UTR of the three target genes were responsible for miR-377 regulation, we cloned the 3′-UTR regions downstream of a luciferase reporter gene and co-transfected these vectors together with control, miR-377 and miR-377-mut into MKN-45 cells, respectively. The luciferase activity of cells transfected with miR-377 was significantly decreased compared with the control. However, the mutation of miR-377 clearly abrogated the repression of the luciferase activity (Fig. 4B). To investigate whether miR-377 regulated these targets, the protein expression levels of p53, PTEN and TIMP1 were assessed by western blot analysis. As shown in Fig. 4C, the overexpression of miR-377 markedly altered the protein levels of p53, PTEN and TIMP1, whereas the protein expression of these target proteins was not affected by mutant miR-377. These results suggested that p53, PTEN and TIMP1 are direct targets of miR-377 and these three targets may mediate the promotive effect of miR-377 on tumorigenesis.

Discussion

GC is one of the most common and lethal cancers, with a high relapse rate (1,14). Therefore, it is essential to develop novel, prognostic factors and therapeutic strategies. The outcome of GC patients is determined primarily by the presence or absence of metastasis (15). Thus, identification of the precise molecular mechanisms that modulate malignant transformation is necessary. Identification of miRNA molecular profiles associated with the prognosis of patients with GC only shed light on the elucidation of the underlying biological mechanisms involved in the development or progression of the disease, and provide the opportunity to identify novel targets for GC diagnosis and treatment (16). To the best of our knowledge, we have shown for the first time that miR-377 was frequently upregulated in GC tissues than in their normal adjacent tissues. Furthermore, the results suggest that miR-377 is involved in the progression of GC. Of note, we found that a high expression of miR-377 was a significant predictor of OS and TTR.

Previous findings suggested that miR-377 plays a vital role in stress responses during pathological conditions, such as diabetes and aging (9,10,17). Overexpression of miR-377 has been observed in human mesangial cells, and was associated with an increased expression of the matrix protein, fibronectin, which is accumulated in excess in diabetic nephropathy (10). In the present study, we first confirmed that miR-377 was frequently upregulated in GC tissues than in their normal adjacent mucosa, and identified a high miR-377 expression as a valid factor associated with advanced tumor stages. These data suggest the potential of miR-377 to serve as a molecular target for GC therapy, especially for tumors with a high potency of metastasis.

The correlation between the miR-377 expression level and clinicopathological values of GC remains unclear. In the present study, we first investigated the relationship of miR-377 expression with clinicopathological characteristics and prognosis. The miR-377 expression was found to be associated with distant metastasis, TNM stage and early TTR, suggesting that miR-377 was involved in the tumorigenesis, development, progression and metastasis of GC. Of note, miR-377 expression was significantly associated with the OS and TTR of GC patients. In support of this, the Kaplan-Meier analysis of overall survival showed that patients whose tumors with a higher miR-377 expression had a significantly worse OS and TTR, indicating that a high miR-377 level is a biomarker of poor prognosis for GC patients. Moreover, the Cox proportional hazards model showed that miR-377 was an independent marker of poor OS and TTR of the known clinical prognostic indicators including distant metastasis, TNM stage and early TTR. Therefore, constitutes a molecular prognostic marker for GC patients, identifying higher risk of death in patients. Thus, good candidates are to receive a more aggressive treatment. To the best of our knowledge, this is the first report to describe the clinical significance of miR-377 in TTR and prognosis of GC patients.

To examine the significance of miR-377 upregulation in GC, several assays were performed. First, we conducted a cell growth assay on MKN-45 cells to assess whether the differential expression of miR-377 affected proliferation. We found that the upregulation of miR-377 expression significantly promoted proliferation and that inhibition of miR-377 suppressed cell proliferation. When assessing the cell cycle of MKN-45 cells by flow cytometry, it was found that miR-377-mimic-transfected cells yielded a population with more cells in the S-phase, and fewer cells in G1-phase, and that inhibition of miR-377 led to cell cycle arrest. The S-phase is the period in which a cell performs DNA replication and the G1/S transition is a major checkpoint in the regulation of the cell cycle (18). Our data suggest that miR-377 affects mechanisms that control the G1/S transition in GC cells.

To investigate the downstream effects of miR-377 overexpression in GC cells, we developed a luciferase-reporter system to assess whether miR-377 can regulate known tumor suppressors and found that miR-377 targets p53, PTEN and TIMP1. p53 is an important tumor-suppressor gene and responds to diverse cellular stresses to regulate the expression of target genes, especially inducing cell cycle arrest and apoptosis (19). Mutations in p53 are associated with a variety of human cancers (20). The p53 has been previously reported to be downregulated in GC tissues, and restoring its expression inhibits growth and reduces clonogenic activity in GC cell lines (21). The result is consistent with our observations that MKN-45 cells have a downregulated p53 expression caused by miR-377, and increased proliferation ability. PTEN has been identified as a tumor suppressor that is mutated in a large number of cancers at high frequency (22). The protein encoding this gene is phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase (23). It negatively regulates the intracellular levels of phosphatidylinositol-3,4,5-trisphosphate in cells and negatively regulates the AKT/PKB signaling pathway (24). Thus, it seems likely that our observation of increased cell numbers in the S-phase and decreased numbers in the G1-phase was caused by the modulation of PTEN by miR-377. TIMP1 has been demonstrated to be a suppressor of tumorigenesis and progression (25). It is a natural inhibitor of the matrix metalloproteinases (MMPs), which are able to promote cell proliferation and may also have an anti-apoptotic function (26). Dysregulation of TIMP1 has been found in several types of cancer (27). In the present study, TIMP1 was found to be downregulated by miR-377 and when miR-377-overexpressing cells exhibited increased growth activity. This result suggests that the dysregulation of TIMP1 by miR-377 is a mechanism associated with this process. The present study provides confounding evidence that an upregulated expression of miR-377 in GC modulates core mechanisms in the control of cell cycle progression and proliferation, by inhibiting the expression of tumor suppressor p53, PTEN and TIMP1. The differential expression of miR-377 in GC can potentially be utilized in diagnostic applications and therapeutic interventions.

In conclusion, to the best of our knowledge, the results of the present study have, for the first time, demonstrated that miR-377 was overexpressed in GC tissue and cell lines and associated with tumorigenesis and poor prognosis. The present study also demonstrated that miR-377 was an independent prognostic factor of patients with GC. Of note, the upregulated expression of miR-377 in GC modulates core mechanisms in the control of proliferation and cell cycle progression, by inhibiting expression of tumor suppressor p53, PTEN and TIMP1. The differential expression of miR-377 in GC can be a candidate therapeutic target and a potential biomarker for the diagnosis and prognosis in GC.

Acknowledgments

The present study was supported by grants from the Agency of Jiangsu Province Science and Technology (no. 2013035) and the Research Office of Jiangsu Cancer Hospital (no. ZK201401).

References

1 

Gibson CJ, Britton KA, Miller AL and Loscalzo J: Clinical problem-solving. Out of the blue. N Engl J Med. 370:1742–1748. 2014.PubMed/NCBI

2 

Wu WKK, Lee CW, Cho CH, Fan D, Wu K, Yu J and Sung JJ: MicroRNA dysregulation in gastric cancer: A new player enters the game. Oncogene. 29:5761–5771. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Song JH and Meltzer SJ: MicroRNAs in pathogenesis, diagnosis, and treatment of gastroesophageal cancers. Gastroenterology. 143:35–47. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Lin X, Yang M, Xia T and Guo J: Increased expression of long noncoding RNA ABHD11-AS1 in gastric cancer and its clinical significance. Med Oncol. 31:422014. View Article : Google Scholar : PubMed/NCBI

5 

Shen L, Shan YS, Hu HM, Price TJ, Sirohi B, Yeh KH, Yang YH, Sano T, Yang HK, Zhang X, et al: Management of gastric cancer in Asia: Resource-stratified guidelines. Lancet Oncol. 14:e535–e547. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Chitwood DH and Timmermans MC: Small RNAs are on the move. Nature. 467:415–419. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Morris KV, Chan SW, Jacobsen SE and Looney DJ: Small interfering RNA-induced transcriptional gene silencing in human cells. Science. 305:1289–1292. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Cai H, Yuan Y, Hao YF, Guo TK, Wei X and Zhang YM: Plasma microRNAs serve as novel potential biomarkers for early detection of gastric cancer. Med Oncol. 30:4522013. View Article : Google Scholar : PubMed/NCBI

9 

Yu X, Luo L, Wu Y, Yu X, Liu Y, Yu X, Zhao X, Zhang X, Cui L, Ye G, et al: Gastric juice miR-129 as a potential biomarker for screening gastric cancer. Med Oncol. 30:3652013. View Article : Google Scholar : PubMed/NCBI

10 

Wang Q, Wang Y, Minto AW, Wang J, Shi Q, Li X and Quigg RJ: MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy. FAsEB J. 22:4126–4135. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Wen ZL, Huang W, Feng YL, Wang Y, Liang J, Cai W, Kang K, Chang D, Zhu P, Milliard RW, et al: MicroRNA-377 regulates angiogenesis by targeting vEGF: Implications for mesenchymal stem cells based therapy in ischemic heart disease. Circulation. 128. 2013

12 

Li N, Liu Q, Su Q, Wei C, Lan B, Wang J, Bao G, Yan F, Yu Y, Peng B, et al: Effects of legumain as a potential prognostic factor on gastric cancers. Med Oncol. 30:6212013. View Article : Google Scholar : PubMed/NCBI

13 

Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A, et al: Treatment of gastric cancer. World J Gastroenterol. 20:1635–1649. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Vicentini C, Fassan M, D’Angelo E, Corbo V, Silvestris N, Nuovo GJ and Scarpa A: Clinical application of microRNA testing in neuroendocrine tumors of the gastrointestinal tract. Molecules. 19:2458–2468. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Beckman JD, Chen C, Nguyen J, Thayanithy V, Subramanian V, Steer CJ and Vercellotti GM: Regulation of heme oxygenase-1 protein expression by miR-377 in combination with miR-217. J Biol Chem. 286:3194–3202. 2011. View Article : Google Scholar :

16 

Zhang R, Luo H, Wang S, Chen W, Chen Z, Wang HW, Chen Y, Yang J, Zhang X, Wu W, et al: MicroRNA-377 inhibited prolife ration and invasion of human glioblastoma cells by directly targeting specificity protein 1. Neuro Oncol. 16:1510–1522. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Tsirimonaki E, Fedonidis C, Pneumaticos SG, Tragas AA, Michalopoulos I and Mangoura D: PKCε signalling activates ERK1/2, and regulates aggrecan, ADAMTs5, and miR377 gene expression in human nucleus pulposus cells. PLoS One. 8:e820452013. View Article : Google Scholar

18 

Bertoli C, Skotheim JM and de Bruin RAM: Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol. 14:518–528. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Duffy MJ, Synnott NC, McGowan PM, Crown J, O’Connor D and Gallagher WM: p53 as a target for the treatment of cancer. Cancer Treat Rev. 40:1153–1160. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Shetzer Y, Solomon H, Koifman G, Molchadsky A, Horesh S and Rotter V: The paradigm of mutant p53-expressing cancer stem cells and drug resistance. Carcinogenesis. 35:1196–1208. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Kubota E, Williamson CT, Ye R, Elegbede A, Peterson L, Lees-Miller SP and Bebb DG: Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle. 13:2129–2137. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Song MS, Salmena L and Pandolfi PP: The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol. 13:283–296. 2012.PubMed/NCBI

23 

Hollander MC, Blumenthal GM and Dennis PA: PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat Rev Cancer. 11:289–301. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Carnero A, Blanco-Aparicio C, Renner O, Link W and Leal JFM: The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets. 8:187–198. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Bjerre C, Vinther L, Belling KC, Würtz SØ, Yadav R, Lademann U, Rigina O, Do KN, Ditzel HJ, Lykkesfeldt AE, et al: TIMP1 overexpression mediates resistance of MCF-7 human breast cancer cells to fulvestrant and down-regulates progesterone receptor expression. Tumour Biol. 34:3839–3851. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Lu YC, Chang JT, Liao CT, Kang CJ, Huang SF, Chen IH, Huang CC, Huang YC, Chen WH, Tsai CY, et al: OncomiR-196 promotes an invasive phenotype in oral cancer through the NME4-JNK-TIMP1-MMP signaling pathway. Mol Cancer. 13:2182014. View Article : Google Scholar : PubMed/NCBI

27 

Bunatova K, Pesta M, Kulda V, Topolcan O, Vrzalova J, Sutnar A, Treska V, Pecen L and Liska V: Plasma TIMP1 level is a prognostic factor in patients with liver metastases. Anticancer Res. 32:4601–4606. 2012.PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 34 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wen X, Wu J, Peng W, Feng J and Tang J: MicroRNA-377 predicts poor clinical outcome of gastric cancer and induces tumorigenesis by targeting multiple tumor-suppressor genes. Oncol Rep 34: 203-210, 2015
APA
Wen, X., Wu, J., Peng, W., Feng, J., & Tang, J. (2015). MicroRNA-377 predicts poor clinical outcome of gastric cancer and induces tumorigenesis by targeting multiple tumor-suppressor genes. Oncology Reports, 34, 203-210. https://doi.org/10.3892/or.2015.3981
MLA
Wen, X., Wu, J., Peng, W., Feng, J., Tang, J."MicroRNA-377 predicts poor clinical outcome of gastric cancer and induces tumorigenesis by targeting multiple tumor-suppressor genes". Oncology Reports 34.1 (2015): 203-210.
Chicago
Wen, X., Wu, J., Peng, W., Feng, J., Tang, J."MicroRNA-377 predicts poor clinical outcome of gastric cancer and induces tumorigenesis by targeting multiple tumor-suppressor genes". Oncology Reports 34, no. 1 (2015): 203-210. https://doi.org/10.3892/or.2015.3981